Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Timothy P. Kegelman is active.

Publication


Featured researches published by Timothy P. Kegelman.


Journal of Cellular Physiology | 2011

Role of Excitatory Amino Acid Transporter-2 (EAAT2) and glutamate in neurodegeneration: Opportunities for developing novel therapeutics

Keetae Kim; Seok-Geun Lee; Timothy P. Kegelman; Zhao-zhong Su; Swadesh K. Das; Rupesh Dash; Santanu Dasgupta; Paola M. Barral; Michael Hedvat; Paul Diaz; John C. Reed; John L. Stebbins; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Glutamate is an essential excitatory neurotransmitter regulating brain functions. Excitatory amino acid transporter (EAAT)‐2 is one of the major glutamate transporters expressed predominantly in astroglial cells and is responsible for 90% of total glutamate uptake. Glutamate transporters tightly regulate glutamate concentration in the synaptic cleft. Dysfunction of EAAT2 and accumulation of excessive extracellular glutamate has been implicated in the development of several neurodegenerative diseases including Alzheimers disease, Huntingtons disease, and amyotrophic lateral sclerosis. Analysis of the 2.5 kb human EAAT2 promoter showed that NF‐κB is an important regulator of EAAT2 expression in astrocytes. Screening of approximately 1,040 FDA‐approved compounds and nutritionals led to the discovery that many β‐lactam antibiotics are transcriptional activators of EAAT2 resulting in increased EAAT2 protein levels. Treatment of animals with ceftriaxone (CEF), a β‐lactam antibiotic, led to an increase of EAAT2 expression and glutamate transport activity in the brain. CEF has neuroprotective effects in both in vitro and in vivo models based on its ability to inhibit neuronal cell death by preventing glutamate excitotoxicity. CEF increases EAAT2 transcription in primary human fetal astrocytes through the NF‐κB signaling pathway. The NF‐κB binding site at −272 position was critical in CEF‐mediated EAAT2 protein induction. These studies emphasize the importance of transcriptional regulation in controlling glutamate levels in the brain. They also emphasize the potential utility of the EAAT2 promoter for developing both low and high throughput screening assays to identify novel small molecule regulators of glutamate transport with potential to ameliorate pathological changes occurring during and causing neurodegeneration. J. Cell. Physiol. 226: 2484–2493, 2011.


Proceedings of the National Academy of Sciences of the United States of America | 2010

Astrocyte elevated gene-1 induces protective autophagy

Sujit K. Bhutia; Timothy P. Kegelman; Swadesh K. Das; Belal Azab; Zhao-zhong Su; Seok-Geun Lee; Devanand Sarkar; Paul B. Fisher

Astrocyte-elevated gene-1 (AEG-1) expression increases in multiple cancers and plays a crucial role in oncogenic transformation and angiogenesis, which are essential components in tumor cell development, growth, and progression to metastasis. Moreover, AEG-1 directly contributes to resistance to chemotherapeutic drugs, another important hallmark of aggressive cancers. In the present study, we document that AEG-1 mediates protective autophagy, an important regulator of cancer survival under metabolic stress and resistance to apoptosis, which may underlie its significant cancer-promoting properties. AEG-1 induces noncanonical autophagy involving an increase in expression of ATG5. AEG-1 decreases the ATP/AMP ratio, resulting in diminished cellular metabolism and activation of AMP kinase, which induces AMPK/mammalian target of rapamycin-dependent autophagy. Inhibition of AMPK by siAMPK or compound C decreases expression of ATG5, ultimately attenuating AEG-1–induced autophagy. AEG-1 protects normal cells from serum starvation-induced death through protective autophagy, and inhibition of AEG-1–induced autophagy results in serum starvation-induced cell death. We also show that AEG-1–mediated chemoresistance is because of protective autophagy and inhibition of AEG-1 results in a decrease in protective autophagy and chemosensitization of cancer cells. In summary, the present study reveals a previously unknown aspect of AEG-1 function by identifying it as a potential regulator of protective autophagy, an important feature of AEG-1 that may contribute to its tumor-promoting properties.


Cancer Research | 2011

Oncogene AEG-1 Promotes Glioma-Induced Neurodegeneration by Increasing Glutamate Excitotoxicity

Seok-Geun Lee; Keetae Kim; Timothy P. Kegelman; Rupesh Dash; Swadesh K. Das; Jung Kyoung Choi; Luni Emdad; Eric L. Howlett; Hyun Yong Jeon; Zhao Zhong Su; Byoung Kwon Yoo; Devanand Sarkar; Sung-Hoon Kim; Dong-Chul Kang; Paul B. Fisher

Aggressive tumor growth, diffuse tissue invasion, and neurodegeneration are hallmarks of malignant glioma. Although glutamate excitotoxicity is considered to play a key role in glioma-induced neurodegeneration, the mechanism(s) controlling this process is poorly understood. Astrocyte elevated gene-1 (AEG-1) is an oncogene that is overexpressed in several types of human cancers, including more than 90% of brain tumors. In addition, AEG-1 promotes gliomagenesis, particularly in the context of tumor growth and invasion, 2 primary characteristics of glioma. In the present study, we investigated the contribution of AEG-1 to glioma-induced neurodegeneration. Pearson correlation coefficient analysis in normal brain tissues and samples from glioma patients indicated a strong negative correlation between expression of AEG-1 and a primary glutamate transporter of astrocytes EAAT2. Gain- and loss-of-function studies in normal primary human fetal astrocytes and T98G glioblastoma multiforme cells revealed that AEG-1 repressed EAAT2 expression at a transcriptional level by inducing YY1 activity to inhibit CBP function as a coactivator on the EAAT2 promoter. In addition, AEG-1-mediated EAAT2 repression caused a reduction of glutamate uptake by glial cells, resulting in induction of neuronal cell death. These findings were also confirmed in samples from glioma patients showing that AEG-1 expression negatively correlated with NeuN expression. Taken together, our findings suggest that AEG-1 contributes to glioma-induced neurodegeneration, a hallmark of this fatal tumor, through regulation of EAAT2 expression.


Cancer Research | 2013

MDA-9/Syntenin and IGFBP-2 Promote Angiogenesis in Human Melanoma

Swadesh K. Das; Sujit K. Bhutia; Belal Azab; Timothy P. Kegelman; Leyla Peachy; Prasanna K. Santhekadur; Santanu Dasgupta; Rupesh Dash; Paul Dent; Steven Grant; Luni Emdad; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Melanoma differentiation-associated gene-9 (mda-9/syntenin) encodes an adapter scaffold protein whose expression correlates with and mediates melanoma progression and metastasis. Tumor angiogenesis represents an integral component of cancer metastasis prompting us to investigate a possible role of mda-9/syntenin in inducing angiogenesis. Genetic (gain-of-function and loss-of-function) and pharmacologic approaches were used to modify mda-9/syntenin expression in normal immortal melanocytes, early radial growth phase melanoma, and metastatic melanoma cells. The consequence of modifying mda-9/syntenin expression on angiogenesis was evaluated using both in vitro and in vivo assays, including tube formation assays using human vascular endothelial cells, chorioallantoic membrane (CAM) assays and xenograft tumor animal models. Gain-of-function and loss-of-function experiments confirm that MDA-9/syntenin induces angiogenesis by augmenting expression of several proangiogenic factors/genes. Experimental evidence is provided for a model of angiogenesis induction by MDA-9/syntenin in which MDA-9/syntenin interacts with the extracellular matrix (ECM), activating Src and FAK resulting in activation by phosphorylation of Akt, which induces hypoxia inducible factor 1-α (HIF-1α). The HIF-1α activates transcription of insulin growth factor-binding protein-2 (IGFBP-2), which is secreted thereby promoting angiogenesis and further induces endothelial cells to produce and secrete VEGF-A augmenting tumor angiogenesis. Our studies delineate an unanticipated cell nonautonomous function of MDA-9/syntenin in the context of angiogenesis, which may directly contribute to its metastasis-promoting properties. As a result, targeting MDA-9/syntenin or its downstream-regulated molecules may provide a means of simultaneously impeding metastasis by both directly inhibiting tumor cell transformed properties (autonomous) and indirectly by blocking angiogenesis (nonautonomous).


Frontiers in Bioscience | 2012

MDA-9/syntenin: a positive gatekeeper of melanoma metastasis.

Swadesh K. Das; Sujit K. Bhutia; Timothy P. Kegelman; Peachy L; Regina A. Oyesanya; Santanu Dasgupta; Upneet K. Sokhi; Belal Azab; Rupesh Dash; Bridget A. Quinn; Keetae Kim; Barral Pm; Zhao-zhong Su; Habib Boukerche; Devanand Sarkar; Paul B. Fisher

Melanoma differentiation associated gene-9 (MDA-9), synonymous with syntenin, is an adapter protein that provides a central role in regulating cell-cell and cell-matrix adhesion. MDA-9/syntenin transduces signals from the cell-surface to the interior through its interaction with a plethora of additional proteins and actively participates in intracellular trafficking and cell-surface targeting, synaptic transmission, and axonal outgrowth. Recent studies demarcate a seminal role of MDA-9/syntenin in cancer metastasis. In the context of melanoma, MDA-9/syntenin functions as a positive regulator of melanoma progression and metastasis through interactions with c-Src and promotes the formation of an active FAK/c-Src signaling complex leading to NF-k B and matrix metalloproteinase (MMP) activation. The present review provides a current perspective of our understanding of the important features of MDA-9/syntenin and its significant role in tumor cell metastasis with special focus on molecular mechanism of action.


Cancer Research | 2014

Astrocyte Elevated Gene-1 Interacts with Akt Isoform 2 to Control Glioma Growth, Survival, and Pathogenesis

Bin Hu; Luni Emdad; Manny D. Bacolod; Timothy P. Kegelman; Xue-Ning Shen; Mohammad A. Alzubi; Swadesh K. Das; Devanand Sarkar; Paul B. Fisher

The oncogene astrocyte elevated gene-1 (AEG-1; MTDH) is highly expressed in glioblastoma multiforme (GBM) and many other types of cancer, where it activates multiple signaling pathways that drive proliferation, invasion, angiogenesis, chemoresistance, radioresistance, and metastasis. AEG-1 activates the Akt signaling pathway and Akt and c-Myc are positive regulators of AEG-1 transcription, generating a positive feedback loop between AEG-1 and Akt in regulating tumorigenesis. Here, we describe in GBM cells a direct interaction between an internal domain of AEG-1 and the PH domain of Akt2, a major driver in GBM. Expression and interaction of AEG-1 and Akt2 are elevated in GBM and contribute to tumor cell survival, proliferation, and invasion. Clinically, in silico gene expression and immunohistochemical analyses of patient specimens showed that AEG-1 and Akt2 expression correlated with GBM progression and reduced patient survival. AEG-1-Akt2 interaction prolonged stabilization of Akt2 phosphorylation at S474, regulating downstream signaling cascades that enable cell proliferation and survival. Disrupting AEG-1-Akt2 interaction by competitive binding of the Akt2-PH domain led to reduced cell viability and invasion. When combined with AEG-1 silencing, conditional expression of Akt2-PH markedly increased survival in an orthotopic mouse model of human GBM. Our study uncovers a novel molecular mechanism by which AEG-1 augments glioma progression and offers a rationale to block AEG-1-Akt2 signaling function as a novel GBM treatment.


Journal of Cellular Physiology | 2012

mda‐7/IL‐24 Differentially Regulates Soluble and Nuclear Clusterin in Prostate Cancer

Sujit K. Bhutia; Swadesh K. Das; Timothy P. Kegelman; Belal Azab; Rupesh Dash; Zhao-zhong Su; Xiang-Yang Wang; Federica Rizzi; Saverio Bettuzzi; Seok-Geun Lee; Paul Dent; Steven Grant; David T. Curiel; Devanand Sarkar; Paul B. Fisher

Melanoma differentiation‐associated gene‐7/interleukin‐24 (mda‐7/IL‐24), a unique member of the IL‐10 gene family, displays a broad range of antitumor properties including cancer‐specific induction of apoptosis, inhibition of tumor angiogenesis, and modulation of anti‐tumor immune responses. Here, we identify clusterin (CLU) as a MDA‐7/IL‐24 interacting protein in DU‐145 cells and investigate the role of MDA‐7/IL‐24 in regulating CLU expression and mediating the antitumor properties of mda‐7/IL‐24 in prostate cancer. Ad.mda‐7 decreased expression of soluble CLU (sCLU) and increased expression of nuclear CLU (nCLU). In the initial phase of Ad.mda‐7 infection sCLU expression increased and CLU interacted with MDA‐7/IL‐24 producing a cytoprotective effect. Infection of stable clones of DU‐145 prostate cancer cells expressing sCLU with Ad.mda‐7 resulted in generation of nCLU that correlated with decreased cell viability and increased apoptosis. In the presence of mda‐7/IL‐24, sCLU‐DU‐145 cells displayed G2/M phase arrest followed by apoptosis. Similarly, Ad.mda‐7 infection decreased cell migration by altering cytoskeleton in sCLU‐DU‐145 cells. Ad.mda‐7‐treated sCLU‐DU‐145 cells displayed a significant reduction in tumor growth in mouse xenograft models and reduced angiogenesis when compared to the vector control group. Tumor tissue lysates demonstrated enhanced nCLU generated from sCLU with increased apoptosis in the presence of MDA‐7/IL‐24. Our findings reveal novel aspects relative to the role of sCLU/nCLU in regulating the anticancer properties of MDA‐7/IL‐24 that may be exploited for developing enhanced therapies for prostate cancer. J. Cell. Physiol. 227: 1805–1813, 2012.


Neuro-oncology | 2014

MDA-9/syntenin is a key regulator of glioma pathogenesis

Timothy P. Kegelman; Swadesh K. Das; Bin Hu; Manny D. Bacolod; Christine E. Fuller; Mitchell E. Menezes; Luni Emdad; Santanu Dasgupta; Albert S. Baldwin; Jeffrey N. Bruce; Paul Dent; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

BACKGROUND The extraordinary invasiveness of human glioblastoma multiforme (GBM) contributes to treatment failure and the grim prognosis of patients diagnosed with this tumor. Consequently, it is imperative to define further the cellular mechanisms that control GBM invasion and identify promising novel therapeutic targets. Melanoma differentiation associated gene-9 (MDA-9/syntenin) is a highly conserved PDZ domain-containing scaffolding protein that promotes invasion and metastasis in vitro and in vivo in human melanoma models. To determine whether MDA-9/syntenin is a relevant target in GBM, we investigated its expression in tumor samples and involvement in GBM invasion and angiogenesis. MATERIALS We assessed MDA-9/syntenin levels in available databases, patient tumor samples, and human-derived cell lines. Through gain-of-function and loss-of-function studies, we analyzed changes in invasion, angiogenesis, and signaling in vitro. We used orthotopic xenografts with GBM6 cells to demonstrate the role of MDA-9/syntenin in GBM pathogenesis in vivo. RESULTS MDA-9/syntenin expression in high-grade astrocytomas is significantly higher than normal tissue counterparts. Forced overexpression of MDA-9/syntenin enhanced Matrigel invasion, while knockdown inhibited invasion, migration, and anchorage-independent growth in soft agar. Moreover, overexpression of MDA-9/syntenin increased activation of c-Src, p38 mitogen-activated protein kinase, and nuclear factor kappa-B, leading to elevated expression of matrix metalloproteinase 2 and secretion of interleukin-8 with corresponding changes observed upon knockdown. GBM6 cells that stably express small hairpin RNA for MDA-9/syntenin formed smaller tumors and had a less invasive phenotype in vivo. CONCLUSIONS Our findings indicate that MDA-9/syntenin is a novel and important mediator of invasion in GBM and a key regulator of pathogenesis, and we identify it as a potential target for anti-invasive treatment in human astrocytoma.


Proceedings of the National Academy of Sciences of the United States of America | 2017

Inhibition of radiation-induced glioblastoma invasion by genetic and pharmacological targeting of MDA-9/Syntenin

Timothy P. Kegelman; Bainan Wu; Swadesh K. Das; Sarmistha Talukdar; Jason M. Beckta; Bin Hu; Luni Emdad; Devanand Sarkar; Frank B. Furnari; Webster K. Cavenee; Jun Wei; Angela Purves; Surya K. De; Maurizio Pellecchia; Paul B. Fisher

Significance In the setting of glioblastoma multiforme (GBM), invasion of cells into normal brain and the unlikeliness of complete surgical removal contributes to GBM lethality and recurrence. “Gold standard” GBM treatment includes adjuvant radiotherapy. Unfortunately, cells surviving radiation demonstrate increased invasion and therapeutic resistance. Melanoma differentiation-associated gene 9 (MDA-9/Syntenin) expression is elevated in patient-derived tumors and GBM cell lines, which correlates with decreased survival and poor response to radiation. Genetic suppression of MDA-9/Syntenin sensitizes GBM to radiation by inhibiting radiation-induced invasion gains and signaling changes. Additionally, intraperitoneal administration of a small-molecule MDA-9/Syntenin inhibitor, PDZ1i, developed using innovative fragment-based drug design and NMR approaches, improved survival of brain tumor-bearing mice. Survival was enhanced further when used with radiation, supporting MDA-9/Syntenin as a therapeutic target for this deadly disease. Glioblastoma multiforme (GBM) is an intractable tumor despite therapeutic advances, principally because of its invasive properties. Radiation is a staple in therapeutic regimens, although cells surviving radiation can become more aggressive and invasive. Subtraction hybridization identified melanoma differentiation-associated gene 9 [MDA-9/Syntenin; syndecan-binding protein (SDCBP)] as a differentially regulated gene associated with aggressive cancer phenotypes in melanoma. MDA-9/Syntenin, a highly conserved double-PDZ domain-containing scaffolding protein, is robustly expressed in human-derived GBM cell lines and patient samples, with expression increasing with tumor grade and correlating with shorter survival times and poorer response to radiotherapy. Knockdown of MDA-9/Syntenin sensitizes GBM cells to radiation, reducing postradiation invasion gains. Radiation induces Src and EGFRvIII signaling, which is abrogated through MDA-9/Syntenin down-regulation. A specific inhibitor of MDA-9/Syntenin activity, PDZ1i (113B7), identified through NMR-guided fragment-based drug design, inhibited MDA-9/Syntenin binding to EGFRvIII, which increased following radiation. Both genetic (shmda-9) and pharmacological (PDZ1i) targeting of MDA-9/Syntenin reduced invasion gains in GBM cells following radiation. Although not affecting normal astrocyte survival when combined with radiation, PDZ1i radiosensitized GBM cells. PDZ1i inhibited crucial GBM signaling involving FAK and mutant EGFR, EGFRvIII, and abrogated gains in secreted proteases, MMP-2 and MMP-9, following radiation. In an in vivo glioma model, PDZ1i resulted in smaller, less invasive tumors and enhanced survival. When combined with radiation, survival gains exceeded radiotherapy alone. MDA-9/Syntenin (SDCBP) provides a direct target for therapy of aggressive cancers such as GBM, and defined small-molecule inhibitors such as PDZ1i hold promise to advance targeted brain cancer therapy.


Expert Opinion on Therapeutic Targets | 2015

Targeting tumor invasion: the roles of MDA-9/Syntenin

Timothy P. Kegelman; Swadesh K. Das; Luni Emdad; Bin Hu; Mitchell E. Menezes; Praveen Bhoopathi; Xiang-Yang Wang; Maurizio Pellecchia; Devanand Sarkar; Paul B. Fisher

Introduction: Melanoma differentiation-associated gene – 9 (MDA-9)/Syntenin has become an increasingly popular focus for investigation in numerous cancertypes. Originally implicated in melanoma metastasis, it has diverse cellular roles and is consistently identified as a regulator of tumor invasion and angiogenesis. As a potential target for inhibiting some of the most lethal aspects of cancer progression, further insight into the function of MDA-9/Syntenin is mandatory. Areas covered: Recent literature and seminal articles were reviewed to summarize the latest collective understanding of MDA-9/Syntenin’s role in normal and cancerous settings. Insights into its participation in developmental processes are included, as is the functional significance of the N- and C-terminals and PDZ domains of MDA-9/Syntenin. Current reports highlight the clinical significance of MDA-9/Syntenin expression level in a variety of cancers, often correlating directly with reduced patient survival. Also presented are assessments of roles of MDA-9/Syntenin in cancer progression as well as its functions as an intracellular adapter molecule. Expert opinion: Multiple studies demonstrate the importance of MDA-9/Syntenin in tumor invasion and progression. Through the use of novel drug design approaches, this protein may provide a worthwhile therapeutic target. As many conventional therapies do not address, or even enhance, tumor invasion, an anti-invasive approach would be a worthwhile addition in cancer therapy.

Collaboration


Dive into the Timothy P. Kegelman's collaboration.

Top Co-Authors

Avatar

Paul B. Fisher

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Swadesh K. Das

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Devanand Sarkar

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Luni Emdad

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Bin Hu

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Santanu Dasgupta

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Rupesh Dash

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Belal Azab

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Mitchell E. Menezes

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge